21 research outputs found

    Cirrose biliar primária e miopatia: uma rara associação

    Get PDF
    Primary biliary cirrhosis (PBC) is a cholestatic liver disease, which is characterized by a chronic inflammatory destruction of intrahepatic bile ducts. It is a rare disorder whose precise etiology is still to be elucidated. Even though the liver is the principal target of PBC, other organ systems also might be affected. Muscular involvement has rarely been described in this disease, and in the majority of cases, muscular weakness has been interpreted as polymyositis. We report the case of a 48-year-old woman suffering from classic PBC, in association with a myopathy whose histological features are distinct from the cases reported before. We also performed a MEDLINE research for PBC and concomitant muscular diseases.A cirrose biliar primária (CBP) é uma doença hepática colestática crônica de etiologia desconhecida e rara. Apesar do principal órgão acometido na CBP ser o fígado, outros órgãos podem também ser afetados. O acometimento muscular raramente tem sido relatado em pacientes com CBP e na maioria destes casos, a fraqueza muscular tem sido interpretada como Polimiosite. Nós relatamos o caso de uma mulher de 48 anos com CBP e miopatia com achados histopatológicos distintos dos casos anteriormente descritos e fizemos uma revisão da literatura sobre o acometimento muscular na CBP

    Activation of Neural and Pluripotent Stem Cell Signatures Correlates with Increased Malignancy in Human Glioma

    Get PDF
    The presence of stem cell characteristics in glioma cells raises the possibility that mechanisms promoting the maintenance and self-renewal of tissue specific stem cells have a similar function in tumor cells. Here we characterized human gliomas of various malignancy grades for the expression of stem cell regulatory proteins. We show that cells in high grade glioma co-express an array of markers defining neural stem cells (NSCs) and that these proteins can fulfill similar functions in tumor cells as in NSCs. However, in contrast to NSCs glioma cells co-express neural proteins together with pluripotent stem cell markers, including the transcription factors Oct4, Sox2, Nanog and Klf4. In line with this finding, in high grade gliomas mesodermal- and endodermal-specific transcription factors were detected together with neural proteins, a combination of lineage markers not normally present in the central nervous system. Persistent presence of pluripotent stem cell traits could only be detected in solid tumors, and observations based on in vitro studies and xenograft transplantations in mice imply that this presence is dependent on the combined activity of intrinsic and extrinsic regulatory cues. Together these results demonstrate a general deregulated expression of neural and pluripotent stem cell traits in malignant human gliomas, and indicate that stem cell regulatory factors may provide significant targets for therapeutic strategies

    Frequent ATRX, CIC, FUBP1 and IDH1 mutations refine the classification of malignant gliomas

    Get PDF
    Mutations in the critical chromatin modifier ATRX and mutations in CIC and FUBP1, which are potent regulators of cell growth, have been discovered in specific subtypes of gliomas, the most common type of primary malignant brain tumors. However, the frequency of these mutations in many subtypes of gliomas, and their association with clinical features of the patients, is poorly understood. Here we analyzed these loci in 363 brain tumors. ATRX is frequently mutated in grade II-III astrocytomas (71%), oligoastrocytomas (68%), and secondary glioblastomas (57%), and ATRX mutations are associated with IDH1 mutations and with an alternative lengthening of telomeres phenotype. CIC and FUBP1 mutations occurred frequently in oligodendrogliomas (46% and 24%, respectively) but rarely in astrocytomas or oligoastrocytomas (<10%). This analysis allowed us to define two highly recurrent genetic signatures in gliomas: IDH1/ATRX (I-A) and IDH1/CIC/FUBP1 (I-CF). Patients with I-CF gliomas had a significantly longer median overall survival (96 months) than patients with I-A gliomas (51 months) and patients with gliomas that did not harbor either signature (13 months). The genetic signatures distinguished clinically distinct groups of oligoastrocytoma patients, which usually present a diagnostic challenge, and were associated with differences in clinical outcome even among individual tumor types. In addition to providing new clues about the genetic alterations underlying gliomas, the results have immediate clinical implications, providing a tripartite genetic signature that can serve as a useful adjunct to conventional glioma classification that may aid in prognosis, treatment selection, and therapeutic trial design.American Cancer Society [RSG-10-126-01-CCE]American Cancer SocietyNCINCI [5R01-CA140316]Pediatric Brain Tumor Foundation Institute GrantPediatric Brain Tumor Foundation Institute GrantSoutheastern Brain Tumor Foundation GrantSoutheastern Brain Tumor Foundation GrantVoices Against Brain Cancer Foundation GrantVoices Against Brain Cancer Foundation GrantJames S. McDonnell Foundation GrantJames S. McDonnell Foundation GrantV FoundationV FoundationAccelerate Brain Cancer Cure Foundation GrantAccelerate Brain Cancer Cure Foundation GrantNIHNIH [5P50 NS20023, 5P50 CA108785, CA057345, CA129825, R37 011898]Sanofi-AventisSanofiAventi

    Correlation of <i>LOX</i>, <i>HIF1A</i> and <i>BMP1</i> expression levels in astrocytomas of different malignant grades.

    No full text
    <p>Correlation was assessed in pilocytic astrocytomas (AGI: A, B and C), low-grade astrocytomas (AGII: D, E and F), anaplastic astrocytomas (AGIII: G, H and I) and glioblastomas (GBM: J, K and L). Statistically significant values were obtained in AGI cases for <i>BMP1</i> and <i>HIF1A</i> correlation (C) and in GBM cases for <i>LOX</i> and <i>BMP1</i> correlation (J), <i>LOX</i> and <i>HIF1A</i> correlation (K) and <i>HIF1A</i> and <i>BMP1</i> expression levels (L). In AGIII samples, <i>HIF1A</i> and <i>LOX</i> expression levels were negatively correlated (H). The statistically significant correlations are shown in red. Correlations between gene expression values were assessed using the non-parametric Spearman-rho correlation test.</p

    LOX expression and localization in astrocytomas and non-neoplastic brain tissues.

    No full text
    <p>Immunohistochemistry was performed in 6 non-neoplastic brain tissues (NN), 6 pilocytic astrocytoma (AGI), 6 low-grade astrocytoma (AGII), 6 anaplastic astrocytoma (AGIII), and 6 glioblastoma (GBM) cases. The images show representative cases of each type of sample (400x magnification for all cases). Arrows indicate the endothelial cells staining.</p

    LOX immunostaining analysis in astrocytomas and non-neoplastic brain tissues.

    No full text
    <p>The graphs illustrate semi-quantitative immunohistochemistry labeling scores (ILS) that refer to the product of staining intensity and the percentage of positive cells stained with anti-LOX in the cytoplasm (A), nucleus (B) and endothelial cells (C). The horizontal bars show the median ILS of each group. The difference in protein expression among the groups was statistically significant for nuclear staining (<i>p</i> = 0.01) and endothelial cell staining (<i>p</i> = 0.0008) as determined by a Kruskal-Wallis test. A post-hoc Dunn’s test was used to calculate the differences in expression among the groups (*<i>p</i><0.05; **<i>p</i><0.001; ***<i>p</i><0.0001).</p

    Effect of <i>LOX</i> knockdown on the invasive phenotype and anchorage-independent growth behavior of GBM cell lines.

    No full text
    <p>Evaluation of the invasion (A and B) and anchorage-independent growth (C and D) of U87MG and A172 cell lines after <i>LOX</i> silencing with siRNA were compared to the non-targeted control (NTC). A total of 2.5 x 10<sup>4</sup> cells were seeded in the upper chamber of Transwell inserts. Cells in the lower chamber were fixed and stained with crystal violet after 18 hours of incubation. Graphs represent the number of migrated cells per field in each condition (means ± standard errors of the means) in the two independent experiments conducted in duplicate. The images show representative fields of U87MG (A) and A172 (B) cells that invaded and crossed the insert (40x magnification). Soft agar colony formation assays were performed for both U87MG (C) and A172 (D) cell lines with 1 x 10<sup>3</sup> cells. Graphs represent the mean number of colonies ± SD of two independent experiments conducted in duplicate (Mann-Whitney test, **<i>p</i><0.001; ***<i>p</i><0.0001).</p

    Gene expression levels in astrocytomas of all malignant grades relative to non-neoplastic samples.

    No full text
    <p>Transcript levels of <i>LOX</i> (A), <i>BMP1</i> (B), and <i>HIF1A</i> (C) were determined in 19 non-neoplastic brain tissue samples (NN), 23 pilocytic astrocytomas (AGI), 26 low-grade astrocytomas (AGII), 18 anaplastic astrocytomas (AGIII) and 84 glioblastomas (GBM). Total RNA was reverse-transcribed into cDNA and analyzed by quantitative real-time PCR (RT-qPCR) using the SYBR Green method. The differences in expression levels among the groups were statistically significant for <i>LOX</i>, <i>BMP1</i> and <i>HIF1A</i> (Kruskal-Wallis test, <i>p</i><0.0001). A post-hoc Dunn’s test was used to calculate the differences in expression between NN and each astrocytoma group (*<i>p</i><0.05; **<i>p</i><0.001; ***<i>p</i><0.0001). <i>IDH1</i> mutational status was determined for diffusely infiltrating cases and the distribution of <i>LOX</i> (D), <i>BMP1</i> (E) and <i>HIF1A</i> (F) expression levels in <i>IDH1</i> wild-type and mutated background. Horizontal bars show the median of each group. AGII and GBM cases with <i>IDH1</i>-mutated presented lower <i>LOX</i> expression levels when compared to <i>IDH1</i> wild-type cases. AGIII cases with wild-type <i>IDH1</i> presented lower <i>HIF1A</i> expression levels when compared to <i>IDH1</i>-mutated cases (Mann-Whitney test, * <i>p</i> <0.05 and t test **, <i>p</i> <0.005).</p

    LOX Expression and Functional Analysis in Astrocytomas and Impact of <i>IDH1</i> Mutation

    No full text
    <div><p>Lysyl oxidase (LOX) is involved in vital biological processes such as cell motility, cell signaling and gene regulation. Deregulation of this protein can contribute to tumor formation and progression. Although it is known that LOX is involved in invasion, proliferation and tumor migration in other types of tumors, studies of LOX in astrocytomas of different grades are scarce. The purpose of our study was to characterize <i>LOX</i>, <i>BMP1</i> and <i>HIF1A</i> expression by real-time PCR in astrocytomas with WHO grades I to IV compared to non-neoplastic brain tissue. <i>IDH1</i> mutational status was determined by PCR and sequencing. LOX protein expression was also analyzed by immunohistochemistry. LOX functional analyses were performed using siRNA knockdown and the specific inhibitor BAPN in two glioblastoma cell lines. The expression levels of <i>LOX</i>, <i>BMP1</i> and <i>HIF1A</i> were correlated and analyzed according to <i>IDH1</i> mutation status and to the clinical end-point of overall survival of glioblastoma patients. The results demonstrate that increased expression and activity of <i>LOX</i>, <i>BMP1</i> and <i>HIF1A</i> were positively correlated with the malignant grade of astrocytomas. LOX protein expression also increased according to the degree of malignancy, with localization in the cytoplasm and nucleus and staining observed in endothelial cells. Glioblastoma with a mutation in <i>IDH1</i> expressed lower levels of LOX in the nucleus, and <i>IDH1</i>-mutated cases showed lower <i>LOX</i> expression levels when compared to wild-type <i>IDH1</i> cases. <i>LOX</i> knockdown and inhibition by BAPN in U87MG and A172 cell lines affected migration, invasion and soft agar colony formation. Taken together, these results corroborate the role of LOX in the migration, invasion and angiogenesis of astrocytomas. Furthermore, <i>LOX</i> expression is influenced by <i>IDH1</i> mutational status. This work provides new insights for researchers aiming to design targeted therapies to control astrocytomas.</p></div
    corecore